You are on page 1of 6

Cell migration in tumors

Hideki Yamaguchi1, Jeffrey Wyckoff1,2 and John Condeelis1,2

Invasion of cancer cells into surrounding tissue and the (3D) culture systems [5,6]. Moreover, recent advances
vasculature is an initial step in tumor metastasis. This requires in in vivo imaging have yielded new insights into the
chemotactic migration of cancer cells, steered by protrusive migration of carcinoma and host cells in tumors and
activity of the cell membrane and its attachment to the demonstrate that the regulation of cancer cell migration
extracellular matrix. Recent advances in intravital imaging and in vivo is more complicated than had been supposed,
the development of an in vivo invasion assay have provided involving chemoattractants, the extracellular matrix, and
new insights into how cancer cell migration is regulated by signaling interactions with stromal cells [2,7,8].
elements of the local microenvironment, including the
extracellular matrix architecture and other cell types found in In this review, we summarize recent progress uncovering
primary tumors. These results, combined with new findings how mammary carcinoma cells migrate in primary tumors
from in vitro studies, have led to new insights into the molecular and how the migration is regulated by macrophages. We
mechanisms of cell protrusive activity and chemotactic also integrate recent findings on molecular mechanisms of
migration during invasion and metastasis. carcinoma cell and macrophage migration, with emphasis
Addresses on the protrusive structures formed by these cell types
1
Department of Anatomy and Structural Biology that steer the cells toward blood vessels and that facilitate
2
Analytical Imaging Facility, Albert Einstein College of Medicine, invasion and intravasation.
1300 Morris Park Avenue, Bronx, New York 10461, USA

Corresponding author: Condeelis, John (condeeli@aecom.yu.edu) Cancer cell migration on and through the
extracellular matrix in tumors in vivo
Cancer cell migration in primary tumors can be directly
Current Opinion in Cell Biology 2005, 17:559–564 observed by multiphoton microscopy with animals carry-
This review comes from a themed issue on ing green fluorescent protein (GFP)-labeled tumors [7].
Cell-to-cell contact and extracellular matrix In the case of breast tumors, most of the migrating cells are
Edited by Inke Näthke and W James Nelson solitary with an amoeboid morphology [9]. These cells are
often observed moving linearly in association with the
Available online 10th August 2005
extracellular matrix (ECM) fibers (Figure 1a). Given that
0955-0674/$ – see front matter some of the ECM fibers converge onto blood vessels, these
# 2005 Elsevier Ltd. All rights reserved. fibers function as a path for carcinoma cells to migrate
toward blood vessels. Directed migration of cancer cells is
DOI 10.1016/j.ceb.2005.08.002
mediated by chemoattractants diffusing from blood ves-
sels and produced by other cell types. Epidermal growth
factor (EGF) receptor expression and chemotactic beha-
Introduction vior in vitro are well correlated with the ability of mammary
Metastasis — the dissemination of cancer cells from the carcinoma cells to invade and metastasize [3,9]. There-
primary tumor to a distant organ — is the most frequent fore, EGF is thought to be a central chemoattractant for
cause of death for patients with cancer. However, the breast cancer invasion. Once carcinoma cells reach blood
molecular mechanisms of metastasis are poorly under- vessels by chemotactic migration, then they must enter the
stood as a result of their apparent complexity. Cancer cell blood space to travel to distant organs. This process, called
migration and invasion into adjacent tissues and intrava- intravasation, requires carcinoma cells to penetrate the
sation into blood/lymphatic vessels are required for dense basement membrane ECM surrounding blood ves-
metastasis of adenocarcinomas, the most common human sel walls and squeeze through the endothelial cell barrier.
cancers [1,2]. Invasive carcinoma cells acquire a migratory Intravital imaging of carcinoma cells in the intravasation
phenotype associated with increased expression of sev- process demonstrated that these cells extend invadopod-
eral genes involved in cell motility [3,4]. This allows like protrusions that penetrate the barriers of the blood
carcinoma cells to respond to cues from the microenvir- vessel wall within minutes [9] (Figure 1b). This observa-
onment that trigger tumor invasion. Therefore, molecules tion indicates that cancer cells form specialized protrusive
involved in cancer cell migration could be potential structures with an ECM-remodeling activity in vivo.
targets for anti-metastasis therapy. However, most studies
on cell motility have been performed in two-dimensional A paracrine loop between cancer cells and
(2D) culture systems, which limits our understanding of macrophages
mechanisms of cell migration, as cells use different cell Tumor progression and invasion are controlled by cross-
migration strategies in physiological three-dimensional talk between cancer cells and other cell types within the

www.sciencedirect.com Current Opinion in Cell Biology 2005, 17:559–564


560 Cell–to–cell contact and ECM

Figure 1

Cell protrusions and the paracrine loop between carcinoma cells and macrophages in intravasation. (a) Model for intravasation of breast cancer
cells. Carcinoma cells, which acquire a migratory phenotype via changes in gene expression resulting from signals from the microenvironment,
degrade underlying basement membrane and detach from the primary tumor. These cells migrate on extracellular matrix (ECM) fibers toward
blood vessels in response to chemoattractants such as epidermal growth factor (EGF). Once cancer cells reach blood vessels they breach the
basement membrane surrounding blood vessel walls and endothelial cells to intravasate into the blood stream. Tumor-associated macrophages
promote carcinoma cell invasion and intravasation by secreting EGF and remodeling ECM architecture. Carcinoma cells also secrete CSF-1
to recruit macrophages to the site of intravasation. (b) Time-lapse images taken by intravital imaging of a GFP-labeled mammary tumor
showing a carcinoma cell on blood vessel wall as it extends an invadopodium-like protrusion into the blood space (arrowheads). Blood vessels
are seen as non-fluorescent regions. The dotted line indicates the cell outline.

tumor and the surrounding tissue. The presence of tumor- receptor and secrete CSF-1. Thus, these two cell types
associated macrophages in primary tumors has been reciprocally induce each other to migrate [11]. This
associated with increased metastasis [10]. Using a che- paracrine loop was further confirmed by reconstitution
motaxis-based in vivo invasion assay, it was revealed that a of invasion in vitro in a 3D culture system [12]. When
paracrine loop between carcinoma cells and macrophages carcinoma cells were co-cultured with macrophages in 3D
facilitates invasion of carcinoma cells in the primary collagen gels, invasion of both cell types was enhanced in
tumor [11]. In the in vivo invasion assay, when micro- an EGF- and CSF-1-dependent manner. Intravital ima-
needles filled with Matrigel containing EGF were ging also demonstrates that carcinoma cells tend to intra-
inserted into a mammary tumor, not only carcinoma cells vasate into the blood stream through sites on blood vessel
but also macrophages migrated into the needles. When walls where tumor-associated macrophages form clusters
colony-stimulating factor 1 (CSF-1), a chemoattractant for (J Wyckoff, unpublished). As macrophages are able to
macrophages, was used instead of EGF in this assay, the degrade ECM through a specialized structure called
same cell types were collected again. Expression analysis podosome, they may generate a local microenvironment
showed that macrophages express CSF-1 receptor and that is favorable for cancer cells to invade and intravasate
secrete EGF, whereas carcinoma cells express EGF by modifying ECM structures (Figure 1a). Cleavage of

Current Opinion in Cell Biology 2005, 17:559–564 www.sciencedirect.com


Cell migration in tumors Yamaguchi, Wyckoff and Condeelis 561

ECM by matrix proteinases produces bioactive frag- naling/adaptor proteins, membrane remodeling proteins
ments, including some chemoattractants for carcinoma and matrix degrading proteases [20,21,22–26,27]
cells [13]. Therefore, macrophages could also contribute (Figure 2). Invadopodium formation has been observed
to chemotaxis of carcinoma cells by releasing chemotactic only in highly invasive cancer cells and is therefore
peptides through podosome-mediated ECM proteolysis. implicated in tumor cell metastasis. EGF has been shown
to stimulate invadopodium formation in carcinoma cells,
Cell protrusions for cell migration in tumors: indicating that invadopodium formation is regulated by
lamellipodia and invadopodia/podosomes chemotactic and invasive signals [27]. Podosomes are
The initial step in cell migration is the protrusion of the dynamic actin-rich adhesion structures that have remark-
cell membrane. This is driven by localized actin poly- able similarity to invadopodia in molecular composition.
merization [14] and can occur in response to chemotactic Podosomes are formed by monocyte-derived cells, such
signals [15]. Carcinoma cells crawling on ECM fibers in as macrophages, as well as by some non-hematopoietic
primary tumors extend pseudopods that attach to the cells and by transformed fibroblasts [28].
fibers at the migration front [7]. The pseudopods seem
to be functionally equivalent to lamellipodia (sheet-like Key molecules for the formation of podosomes and inva-
protrusions observed in cells migrating on 2D substrate in dopodia are WASP (Wiskott-Aldrich Syndrome protein)
vitro), although the shapes of pseudopods are more three and N-WASP (neural WASP). WASP and N-WASP are
dimensional in vivo [9]. Leading protrusions attach to WASP family proteins that induce the rapid actin poly-
collagen-containing fibers, a process that may involve the merization required for formation of cell protrusions
dynamic adhesion structures called focal complexes. The [29–31]. WASP has been shown to regulate podosome
upregulation in invasive tumor cells of the expression of formation in macrophages, dendritic cells and osteoclasts
adhesion molecules such as laminins and integrins [3,9] [28,32]. N-WASP is a component of podosomes and
is consistent with the importance of integrins in Rac1 invadopodia in non-hematopoietic and cancer cells
stimulation, adhesion formation and cell migration in [27,28,33–35]. Several upstream activators of N-WASP/
invasive tumor cells [16]. WASP, including Cdc42, Nck1 and WIP, and a down-
stream effector, the Arp2/3 complex, have been shown to
The protrusion activity of invasive tumor cells is function in invadopodium/podosome formation [21,
enhanced by their upregulation of the genes responsible 27,28,33,36,37]. Thus, the WASP/N-WASP signaling
for EGF-stimulated protrusions, namely the genes coding pathway is likely to have a role in ECM remodeling via
for the cofilin, capping protein and Arp2/3 complex path- the formation of podosomes and invadopodia and, hence,
ways [3,4]. The significance of this pattern of expression in tumor invasion and metastasis. Supporting this hypoth-
in invasive tumor cells may be understood when one esis, our pilot experiments have shown that mammary
considers that the cofilin pathway is directly involved tumors of the rat derived from carcinoma cells expressing
in directional sensing during chemotaxis of carcinoma dominant-negative N-WASP have a reduced ability to
cells to EGF [17], and that cofilin is sufficient to set the intravasate and spontaneously metastasize (H Yamaguchi,
direction of cell movement [18]. Directional sensing of unpublished).
EGF requires a burst of generation of free, actin filament
barbed ends resulting from cofilin severing that causes Although there are many similarities between invadopo-
localized actin polymerization [17]. If the free barbed end dia and podosomes, their precise relationship has been
formation is either inhibited or sustained, then directional obscured by the fact that most podosome studies have
protrusion in response to EGF fails. The cofilin-depen- used cells on non-physiological surfaces such as glass,
dent actin polymerization has also been shown to act in whereas studies of invadopodia are classically carried out
synergy with the Arp2/3 complex to generate protrusions on thick ECM. Given that this might lead to an artificial
in response to EGF that are responsible for cell migration distinction between the two structures, it has been pro-
[15,19]. posed that podosomes are precursor structures that
mature into more stable invadopodia under certain phy-
To migrate through a physical barrier of dense ECM, siological conditions [21,27]. This is supported by evi-
cancer cells need to extend protrusions with an ability to dence that podosomes show an ECM degradation activity
remodel and degrade ECM. This type of protrusion is on physiological substrates [28,38–40]. Recent studies
particularly important for the invasion of cancer cells using time-lapse microscopy revealed that podosomes are
through the basement membrane covering blood vessels. dynamic structures with a life span of minutes, whereas
Invasive cancer cells cultured on physiological substrates invadopodia have lifetimes that vary from minutes to
form actin-rich membrane protrusions extended verti- hours [27,41,42]. Interestingly, invadopodia with long
cally from the ventral cell membrane. These structures lifetimes are more stationary than short-lived invadopo-
have been termed invadopodia, because they have a dia, suggesting that stable functional invadopodia mature
matrix degradation activity. Invadopodia are enriched from dynamic motile precursors that are conceivably
with actin regulatory proteins, adhesion molecules, sig- equivalent to podosomes. This maturation process

www.sciencedirect.com Current Opinion in Cell Biology 2005, 17:559–564


562 Cell–to–cell contact and ECM

Figure 2

Model of carcinoma cell migration on and through the extracellular matrix (ECM). (a) Highly motile carcinoma cells use dynamic adhesion
structures, focal complexes and podosome-type invadopodia precursors for migration on the ECM. In response to invasive signals, these
structures mature into stable invadopodia when cells migrate through rigid ECM. N-WASP nucleates actin filaments to initiate invadopodium/
podosome formation and also induce actin network formation synergistically with cofilin at the base to anchor and stabilize invadopodia.
(b) The molecular components of invadopodia.

appears to require integrin activation, concentration of Src or Ras disassembles focal adhesions and stress fibers
matrix proteinases, and stabilization via the formation of and induces the formation of podosomes, a process that is
actin network by the actin polymerization machinery. associated with increased cell motility and invasion
FRET (fluorescence resonance energy transfer) analyses [44,45]. Taken together, focal complexes mainly mature
with an N-WASP biosensor demonstrated that N-WASP into stable focal adhesions associated with stress fibers in
is transiently activated at the base of active invadopodia strongly adherent, slow-migratory cells, whereas in highly
[43]. Cofilin, a critical initiator of actin polymerization in motile and invasive cells these structures are converted to
response to EGF, has been shown to determine the site of dynamic podosomes/invadopodia precursors, and can
actin polymerization and, in synergy with Arp2/3 com- subsequently mature into functional invadopodia that
plex, to enhance dendritic nucleation [15]. Cofilin knock- allow cells to invade and degrade ECM barriers
down results in the formation of less invasive invadopodia (Figure 2).
with short lifetimes, indicating that cofilin is required for
maturation of invadopodia [27]. Therefore, N-WASP/ Conclusions
Arp2/3 complex and cofilin seem to cooperatively induce Mechanisms of cell migration in tumors have been
actin polymerization at the base of invadopodia to anchor unveiled by the use of new technologies such as intravital
and stabilize invadopodia (Figure 2). imaging and in vivo invasion assays. In vitro studies using
3D cell culture systems have revealed new aspects of
Macrophage podosomes are formed at the leading lamel- cancer cell migration and focused our attention on spe-
lipodia and are thought to function in adhesion site cialized protrusions involved in chemotaxis and ECM
formation, which is required for chemotaxis [28,42]. invasion. Nevertheless, 2D cell culture still provides
Interestingly, focal adhesions in smooth muscle cells important insights into the molecular basis of cell protru-
are remodeled into podosomes at the cell periphery in sions, chemotaxis and cell polarity. Therefore, the inte-
response to phorbol ester, a potent tumor promoter [37]. gration of in vivo, in vitro, 3D, and 2D observations will be
Given that focal complexes/adhesions and podosomes required to describe mechanisms of cancer cell migration
share major structural components, it is possible that precisely. Integration of these approaches has already led
podosomes are converted from focal complexes/adhe- to a better understanding of the relationships between
sions upon cell migratory signals. This is consistent with and importance of podosomes and invadopodia. Future
the fact that transformation of fibroblasts by oncogenic advances using these combined approaches will hopefully

Current Opinion in Cell Biology 2005, 17:559–564 www.sciencedirect.com


Cell migration in tumors Yamaguchi, Wyckoff and Condeelis 563

lead to the development of a new generation of anti- 14. Pollard TD, Borisy GG: Cellular motility driven by assembly
and disassembly of actin filaments. Cell 2003,
cancer drugs that target cell motility. 112:453-465.
15. DesMarais V, Ghosh M, Eddy R, Condeelis J: Cofilin takes the
Acknowledgements lead. J Cell Sci 2005, 118:19-26.
We apologize to those authors whose work we were unable to cite
because of space and date of publication limitations. We are grateful 16. Yu W, Datta A, Leroy P, O’Brien LE, Mak G, Jou TS, Matlin KS,
Mostov KE, Zegers MM: Beta1-integrin orients epithelial
to all laboratory members and collaborators for their support and helpful
polarity via Rac1 and laminin. Mol Biol Cell 2005,
discussions. We thank M Sidani for intravital imaging pictures. This work 16:433-445.
was supported by grants from the NIH (GM38511 and CA100324).
17. Mouneimne G, Soon L, DesMarais V, Sidani M, Song X,
Yip SC, Ghosh M, Eddy R, Backer JM, Condeelis J:
References and recommended reading Phospholipase C and cofilin are required for carcinoma
Papers of particular interest, published within the annual period of cell directionality in response to EGF stimulation. J Cell Biol
review, have been highlighted as: 2004, 166:697-708.

 of special interest 18. Ghosh M, Song X, Mouneimne G, Sidani M, Lawrence DS,


 of outstanding interest  Condeelis JS: Cofilin promotes actin polymerization and
defines the direction of cell motility. Science 2004,
1. Chambers AF, Groom AC, MacDonald IC: Dissemination and 304:743-746.
growth of cancer cells in metastatic sites. Nat Rev Cancer 2002, A direct demonstration of cofilin function in vivo, revealing cofilin’s ability
2:563-572. to steer the direction of cell locomotion.

2. Friedl P, Wolf K: Tumour-cell invasion and migration: diversity 19. DesMarais V, Macaluso F, Condeelis J, Bailly M: Synergistic
and escape mechanisms. Nat Rev Cancer 2003, 3:362-374. interaction between the Arp2/3 complex and cofilin
drives stimulated lamellipod extension. J Cell Sci 2004,
3. Wang W, Goswami S, Lapidus K, Wells AL, Wyckoff JB, Sahai E, 117:3499-3510.
 Singer RH, Segall JE, Condeelis JS: Identification and testing of
a gene expression signature of invasive carcinoma cells within 20. Baldassarre M, Pompeo A, Beznoussenko G, Castaldi C,
primary mammary tumors. Cancer Res 2004, 64:8585-8594. Cortellino S, McNiven MA, Luini A, Buccione R: Dynamin
This paper describes the first example of an invasion signature for breast participates in focal extracellular matrix degradation by
cancer, which relates invasiveness to pathways driving cell motility. invasive cells. Mol Biol Cell 2003, 14:1074-1084.

4. Wang W, Goswami S, Sahai E, Wyckoff JB, Segall JE, 21. Buccione R, Orth JD, McNiven MA: Foot and mouth:
Condeelis JS: Tumor cells caught in the act of invading: their  podosomes, invadopodia and circular dorsal ruffles.
strategy for enhanced cell motility. Trends Cell Biol 2005, Nat Rev Mol Cell Biol 2004, 5:647-657.
15:138-145. This review summarizes the current knowledge of podosomes, invado-
podia and dorsal ruffles and links the characteristics of these structures to
5. Wolf K, Mazo I, Leung H, Engelke K, von Andrian UH, Deryugina EI, a useful list of their molecular components.
Strongin AY, Brocker EB, Friedl P: Compensation mechanism
in tumor cell migration: mesenchymal-amoeboid transition 22. Chuang YY, Tran NL, Rusk N, Nakada M, Berens ME, Symons M:
after blocking of pericellular proteolysis. J Cell Biol 2003, Role of synaptojanin 2 in glioma cell migration and invasion.
160:267-277. Cancer Res 2004, 64:8271-8275.

6. Sahai E, Marshall CJ: Differing modes of tumour cell invasion 23. Hashimoto S, Onodera Y, Hashimoto A, Tanaka M,
have distinct requirements for Rho/ROCK signalling and Hamaguchi M, Yamada A, Sabe H: Requirement for Arf6 in
extracellular proteolysis. Nat Cell Biol 2003, 5:711-719. breast cancer invasive activities. Proc Natl Acad Sci USA
2004, 101:6647-6652.
7. Condeelis J, Segall JE: Intravital imaging of cell movement in
 tumours. Nat Rev Cancer 2003, 3:921-930. 24. McHugh B, Krause SA, Yu B, Deans AM, Heasman S, McLaughlin
This review describes the mechanisms of carcinoma cell migration in P, Heck MM: Invadolysin: a novel, conserved metalloprotease
primary tumors on the basis of observations obtained by intravital multi- links mitotic structural rearrangements with cell migration.
photon microscopy. Differences in cell migration between in vitro and in J Cell Biol 2004, 167:673-686.
vivo and between metastatic and non-metastatic cells are described.
25. Onodera Y, Hashimoto S, Hashimoto A, Morishige M, Mazaki Y,
8. Sahai E: Mechanisms of cancer cell invasion. Curr Opin Genet Yamada A, Ogawa E, Adachi M, Sakurai T, Manabe T et al.:
Dev 2005, 15:87-96. Expression of AMAP1, an ArfGAP, provides novel targets
to inhibit breast cancer invasive activities. EMBO J 2005,
9. Wang W, Wyckoff JB, Frohlich VC, Oleynikov Y, Huttelmaier S, 24:963-973.
Zavadil J, Cermak L, Bottinger EP, Singer RH, White JG et al.:
Single cell behavior in metastatic primary mammary tumors 26. Tague SE, Muralidharan V, D’Souza-Schorey C: ADP-ribosylation
correlated with gene expression patterns revealed by factor 6 regulates tumor cell invasion through the activation of
molecular profiling. Cancer Res 2002, 62:6278-6288. the MEK/ERK signaling pathway. Proc Natl Acad Sci USA 2004,
101:9671-9676.
10. Pollard JW: Tumour-educated macrophages promote
tumour progression and metastasis. Nat Rev Cancer 2004, 27. Yamaguchi H, Lorenz M, Kempiak S, Sarmiento C, Coniglio S,
4:71-78.  Symons M, Segall J, Eddy R, Miki H, Takenawa T et al.: Molecular
mechanisms of invadopodium formation: the role of the
11. Wyckoff J, Wang W, Lin EY, Wang Y, Pixley F, Stanley ER, N-WASP-Arp2/3 complex pathway and cofilin. J Cell Biol 2005,
 Graf T, Pollard JW, Segall J, Condeelis J: A paracrine loop 168:441-452.
between tumor cells and macrophages is required for This study demonstrates that functional invadopodia are derived from
tumor cell migration in mammary tumors. Cancer Res 2004, dynamic precursors and also shows the importance of the N-WASP/
64:7022-7029. Arp2/3 complex pathway and cofilin in invadopodium formation.
Using an in vivo invasion assay, this study reveals the paracrine signaling
mechanisms between microphages and carcinoma cells in cancer inva- 28. Linder S, Aepfelbacher M: Podosomes: adhesion hot-spots of
sion involving EGF-receptor- and CSF-1-receptor-mediated cell motility.  invasive cells. Trends Cell Biol 2003, 13:376-385.
An important review describing the molecular basis and functional fea-
12. Goswami S, Sahai E, Wyckoff J, Cammer M, Cox D, Pixley F, tures of podosomes.
Stanley ER, Segall J, Condeelis J: Macrophages promote the
invasion of breast carcinoma cells via a CSF-1/EGF paracrine 29. Miki H, Takenawa T: Regulation of actin dynamics by WASP
loop. Cancer Res 2005, 65:5278-5283. family proteins. J Biochem (Tokyo) 2003, 134:309-313.
13. Mott JD, Werb Z: Regulation of matrix biology by matrix 30. Millard TH, Sharp SJ, Machesky LM: Signalling to actin
metalloproteinases. Curr Opin Cell Biol 2004, 16:558-564. assembly via the WASP (Wiskott-Aldrich syndrome protein)

www.sciencedirect.com Current Opinion in Cell Biology 2005, 17:559–564


564 Cell–to–cell contact and ECM

family proteins and the Arp2/3 complex. Biochem J 2004, 38. Berdeaux RL, Diaz B, Kim L, Martin GS: Active Rho is localized to
380:1-17. podosomes induced by oncogenic Src and is required for their
assembly and function. J Cell Biol 2004, 166:317-323.
31. Stradal TE, Rottner K, Disanza A, Confalonieri S, Innocenti M,
Scita G: Regulation of actin dynamics by WASP and WAVE 39. Seals DF, Azucena EF Jr, Pass I, Tesfay L, Gordon R, Woodrow M,
family proteins. Trends Cell Biol 2004, 14:303-311. Resau JH, Courtneidge SA: The adaptor protein Tks5/Fish is
required for podosome formation and function, and for the
32. Calle Y, Chou HC, Thrasher AJ, Jones GE: Wiskott-Aldrich protease-driven invasion of cancer cells. Cancer Cell 2005,
syndrome protein and the cytoskeletal dynamics of dendritic 7:155-165.
cells. J Pathol 2004, 204:460-469.
40. Burgstaller G, Gimona M: Podosome-mediated matrix
33. Moreau V, Tatin F, Varon C, Genot E: Actin can reorganize into
resorption and cell motility in vascular smooth muscle cells.
podosomes in aortic endothelial cells, a process controlled by
Am J Physiol Heart Circ Physiol 2005, 288:H3001-H3005.
Cdc42 and RhoA. Mol Cell Biol 2003, 23:6809-6822.
34. Spinardi L, Rietdorf J, Nitsch L, Bono M, Tacchetti C, Way M, 41. Destaing O, Saltel F, Geminard JC, Jurdic P, Bard F: Podosomes
Marchisio PC: A dynamic podosome-like structure of epithelial display actin turnover and dynamic self-organization in
cells. Exp Cell Res 2004, 295:360-374. osteoclasts expressing actin-green fluorescent protein.
Mol Biol Cell 2003, 14:407-416.
35. Park SJ, Suetsugu S, Takenawa T: Interaction of HSP90 to
N-WASP leads to activation and protection from proteasome- 42. Evans JG, Correia I, Krasavina O, Watson N, Matsudaira P:
dependent degradation. EMBO J 2005, 24:1557-1570. Macrophage podosomes assemble at the leading lamella by
growth and fragmentation. J Cell Biol 2003, 161:697-705.
36. Nakahara H, Otani T, Sasaki T, Miura Y, Takai Y, Kogo M:
Involvement of Cdc42 and Rac small G proteins in invadopodia 43. Lorenz M, Yamaguchi H, Wang Y, Singer RH, Condeelis J:
formation of RPMI7951 cells. Genes Cells 2003, 8:1019-1027. Imaging sites of N-WASP activity in lamellipodia and
invadopodia of carcinoma cells. Curr Biol 2004,
37. Kaverina I, Stradal TE, Gimona M: Podosome formation in 14:697-703.
 cultured A7r5 vascular smooth muscle cells requires
Arp2/3-dependent de novo actin polymerization at discrete 44. Martin GS: Cell signaling and cancer. Cancer Cell 2003,
microdomains. J Cell Sci 2003, 116:4915-4924. 4:167-174.
A demonstration that podosomes are formed at the interface between
stress fibers and focal adhesions upon phorbol ester stimulation in 45. Frame MC: Newest findings on the oldest oncogene; how
smooth muscle cells. activated src does it. J Cell Sci 2004, 117:989-998.

Current Opinion in Cell Biology 2005, 17:559–564 www.sciencedirect.com

You might also like